EVI1 dysregulation: impact on biology and therapy of myeloid malignancies - Nature

Page created by Kim Olson
 
CONTINUE READING
Birdwell et al. Blood Cancer Journal (2021)11:64
                                    https://doi.org/10.1038/s41408-021-00457-9                                                                                                          Blood Cancer Journal

                                     REVIEW ARTICLE                                                                                                                                             Open Access

                                    EVI1 dysregulation: impact on biology and therapy
                                    of myeloid malignancies
                                    Christine Birdwell1, Warren Fiskus                        1
                                                                                                , Tapan M. Kadia               1
                                                                                                                                 , Courtney D. DiNardo1, Christopher P. Mill1 and
                                    Kapil N. Bhalla 1

                                      Abstract
                                      Ecotropic viral integration site 1 (Evi1) was discovered in 1988 as a common site of ecotropic viral integration resulting
                                      in myeloid malignancies in mice. EVI1 is an oncogenic zinc-finger transcription factor whose overexpression
                                      contributes to disease progression and an aggressive phenotype, correlating with poor clinical outcome in myeloid
                                      malignancies. Despite progress in understanding the biology of EVI1 dysregulation, significant improvements in
                                      therapeutic outcome remain elusive. Here, we highlight advances in understanding EVI1 biology and discuss how this
                                      new knowledge informs development of novel therapeutic interventions. EVI1 is overexpression is correlated with
                                      poor outcome in some epithelial cancers. However, the focus of this review is the genetic lesions, biology, and current
                                      therapeutics of myeloid malignancies overexpressing EVI1.

                                    MECOM locus discovery                                                                               EVI1: domain-structure and function
                  1234567890():,;
1234567890():,;
                  1234567890():,;
1234567890():,;

                                       Evi1 was discovered by Mucenski et al. as a common                                                 Human EVI1 is a 145 kilo Dalton (kDa) protein that
                                    site of ecotropic viral integration in mice that caused                                             contains 1051 amino acids. EVI1 localizes to the nucleus
                                    virally induced myeloid malignancies1. Through its rear-                                            and binds DNA through its zinc finger (ZF) domains5.
                                    rangements in human acute myeloid leukemia (AML), the                                               EVI1 contains ten zinc fingers that are arranged in two
                                    human EVI1 gene was mapped to the long arm of chro-                                                 separate sets, one N-terminal containing seven zinc fin-
                                    mosome 3 at q 26.2 (3q26.2)2. The EVI1 gene in humans                                               gers, another C-terminal containing three zinc fingers5
                                    is ~92% homologous to the mouse Evi12. EVI1 is encoded                                              (Fig. 1). Through electrophoretic mobility shift assays and
                                    from the MDS1 and ecotropic viral integration site 1                                                chromatin immunoprecipitation (ChIP) assays, the N-
                                    (EVI1) complex locus (MECOM), which includes several                                                terminal ZF domain was determined to bind TAGA/
                                    alternative transcripts3. EVI1 exists either as a shorter                                           TCTA or CAGAGA/TCTCTG GATA-like simple
                                    single gene or as spliced to the short myelodysplastic                                              sequence repeats (SSRs)5,6. The C-terminal ZF domain
                                    syndrome 1 (MDS1) gene, present more than 350 kb                                                    recognizes a CCATATAA ETS-like motif5,6. In the region
                                    upstream to EVI1, creating the longer MDS1-EVI1 gene3.                                              between the ZF domains, is the repressor region that
                                    The shorter isoform of EVI1 is abundant and onco-                                                   contains the interaction sites for the co-repressor CtBP
                                    genic4,5. A truncated variant of the EVI1 transcript con-                                           (C-terminal binding protein 1)7,8. EVI1 also contains an
                                    served in both mice and humans, EVI1Δ324, lacks part of                                             acidic domain at its C terminus5 (Fig. 1).
                                    the first zinc finger domain and the ability to transform                                               Disruption of the full-length Evi1 transcript by muta-
                                    (Fig. 1)4.                                                                                          genesis in mice led to severe developmental defects in the
                                                                                                                                        heart and central nervous system, and homozygous
                                                                                                                                        mutants died at approximately embryonic day 10.59.
                                                                                                                                        Additionally, adult mice with conditional knockout of
                                                                                                                                        Evi1 had a marked reduction in their long-term hema-
                                    Correspondence: Kapil N. Bhalla (kbhalla@mdanderson.org)
                                    1
                                     Division of Cancer Medicine, Department of Leukemia, The University of Texas                       topoietic stem cells (LT-HSCs), and upon transfer into
                                    M. D. Anderson Cancer Center, Houston, TX 77030, USA

                                    © The Author(s) 2021
                                                        Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction
                                                        in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if
                                    changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If
                                    material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
                                    permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                                    Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                                             Page 2 of 14

                         EVI1                          Zinc Fingers (1-7)                            Zinc Fingers (8-10)
                       (145 kDa)
                                                                                        Repressive                         Acidic
                         1051 aa             NH2-                                        Domain                            Domain    -COOH

                                                1                           239   547                   733       812 886      937

                       MDS1-EVI1                       Zinc Fingers (1-7)                            Zinc Fingers (8-10)
                       (200 kDa)
                                      PR Domain                                         Repressive                         Acidic    -COOH
                               NH2-    (188 aa)                                          Domain                            Domain
                                    PRDF1-RIZ (PR)
                                   homology domain
                                                            Truncated Zinc
                                                            Finger domain
                                                                                                     Zinc Fingers (8-10)
                       EVI1∆324                             1    2 3 4      5
                       (105 kDa)
                                                                                        Repressive                         Acidic
                                                     NH2-                                Domain                                      -COOH
                                                                                                                           Domain

  Fig. 1 Schematic of the MDS and EVI1 (MECOM) locus proteins. C2H2-zinc finger motifs are shown in blue. Zinc finger motifs 1–7 and motifs
  8–10 form the N-terminal and C-terminal zinc finger domains respectively. The repressive domain and acidic domain are depicted in tan and red,
  respectively. The numbers beneath the schematic indicate the amino acid positions of the zinc finger domains, the repressive domain and the acidic
  domain of EVI1.

irradiated mice were unable to engraft and repopulate                             (GATA binding protein 1) and PU.1 (transcription factor
efficiently10. The self-renewal ability of LT-HSCs is linked                       PU.1)17,19. In the megakaryocyte lineage, EVI1 is expres-
to EVI1 expression, and many LT-HSC-associated genes                              sed in early precursor cells20. In a transgenic mouse model
have EVI1 binding sites in their regulatory regions11.                            recapitulating human inv3(q21q26) AML that over-
Furthermore, increased EVI1 expression is a common                                expresses EVI1 and also has GATA2 haploinsufficiency,
immortalizing factor of murine primary bone marrow                                EVI1 and GATA2 dysregulation together skewed hema-
after retroviral infection12. For example, MSCV integra-                          topoiesis toward the megakaryocyte lineage more so than
tion promoted increased expression of EVI1 causing                                EVI1 overexpression alone21. This suggests that EVI1 may
immortalization of immature myeloid cells, but they were                          work in concert with other factors to promote the
unable to induce leukemia in transplanted hosts12. Thus                           megakaryocyte lineage21. In general, for most myeloid
EVI1 supports HSC self-renewal, but EVI1 expression                               lineages, EVI1 functions to promote a stem or early
alone is not enough to drive leukemogenesis12,13.                                 progenitor transcriptional program11,14. Forced EVI1
  In addition to LT-HSC self-renewal, expression of EVI1                          expression maintains the stem-like program while
blocks hematopoietic differentiation of the granulocyte,                          simultaneously suppressing myeloid transcription factors
erythroid, dendritic, and monocytic lineages14,15. EVI1                           involved in myeloid differentiation16,18,19,21. Notably,
expression in primary mouse myeloid progenitor cells                              endogenous EVI1 is generally downregulated under nor-
upregulated HSC-associated genes and decreased DNA                                mal differentiation14,15. However, the degree to which
replication and repair genes14. EVI1 transcripts are                              endogenous EVI1 blocks differentiation and what factors
decreased in human CD34+ cells after stimulation of                               normally downregulate EVI1 during differentiation largely
differentiation induced by cytokine administration, sug-                          remain an unknown.
gesting that downregulation of EVI1 is an important step
in terminal differentiation of many hematopoietic linea-                          MDS1-EVI1 and EVI1Δ324
ges15. Forced expression of Evi1 in the mouse bone                                  In 1994, Nucifora et al. identified a transcript of
marrow cell line 32Dcl3 inhibits differentiation response                         unknown function that they termed MDS1, which formed
to granulocytes and erythrocytes due to granulocyte                               a fusion protein with RUNX1 and/or EVI1 in several
colony-stimulating factor (G-CSF) and erythropoietin,                             myelodysplastic syndrome (MDS) patients22. Currently,
respectively16,17. Extrinsic EVI1 expression blocked G-                           the function of the MDS1 protein itself is still unknown.
CSF-induced differentiation through transcriptional                               In MDS1-EVI1, exon 2 of MDS1 is fused in-frame to EVI1
repression of the lineage-specific gene myeloperoxidase                            exon 2, which adds 188 amino acids upstream of the
and the myeloid transcription factors C/EBPα (CCAAT                               normal start codon of EVI1 in exon 322. A part of these
enhancer binding protein alpha) and RUNX1 (runt-rela-                             extra N-terminal amino acids contains the PR domain,
ted transcription factor 1, also known as AML1)16,18.                             which shares homology with the B cell factor positive
Erythroid differentiation was blocked by EVI1 through                             regulatory domain 1-binding factor (PRD1-BF1) and
binding and subsequent inhibition of transcriptional                              retinoblastoma binding protein RIZ13,12. The PR domain
activity of the myeloid transcription factors GATA1                               is related to a subset of the methyltransferase SET

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                                  Page 3 of 14

                                                       318 bp Minimal Promoter for EVI1

                        GATA1         MYB            GATA1            ELK1          MYB   RUNX1        PU.1       EVI1
                                                     GATA2

                                                                                                   (TTCTCCTCCTT   Transcription
                       (GGCGATGT) (CACCGTTTCT) (CTGCTTATCTACGT) (GCGATTTCC) (CGAAACGG) (TGCGGTC)
                                                                                                    CCCCTCCCTC)   Factor Motifs

  Fig. 2 Schematic representation of the 318-bp minimal promoter of EVI1 in humans. Within the upstream EVI1 promoter, the positions and
  nucleotide sequences/binding sites of specific transcription factors are shown.

domains3,23 (Fig. 1). The combination of a PR domain and                     binding motifs for RUNX1, ELK1 (ETS transcription
zinc finger domains in MDS1-EVI1 makes it a part of the                       factor ELK1), RELA (RELA proto-oncogene, NF-κB
PRDM (PR/SET domain) family and thus is also called                          Subunit), GATA1, and MYB (MYB proto-oncogene,
PRDM3, which was characterized as a mono-methyl                              transcription factor). Knockdown of RUNX1 and/or ELK1
H3K9 methyltransferase23. Although the specific role of                       in HEL cells decreased EVI1 mRNA and protein levels26.
MDS1-EVI1 is not always separated from the role of EVI1,                     Furthermore, interactions between RUNX1 and EVI1 at
loss of MDS1-EVI1 is also associated with embryonic                          the minimal promoter appear to positively regulate EVI1
lethality, developmental defects, and dysregulation of                       activity26. MDS1-EVI1 and EVI1Δ324 bind further
hematopoiesis10,15.                                                          downstream of the minimal promoter of EVI1 and reduce
  EVI1Δ324 is a variant transcript of EVI1 with an                           its transcription26,27. MDS1-EVI1 and EVI1Δ324 are
internal 972 nucleotide deletion that removes the 6th and                    reported to be co-expressed with EVI115,24. Although not
7th zinc finger from the N-terminal ZF domain4 (Fig. 1).                      further studied, present upstream of the minimal EVI1
ChIP assays with FLAG-tagged EVI1 or EVI1Δ324 in an                          promoter are the consensus binding motifs for GATA1,
ovarian carcinoma cell line (SKOV3) showed an ~71%                           GATA2, and C/EBPα, suggesting that MDS1-EVI1 and
overlap in binding peaks between the two24. Additionally,                    EVI1Δ324 may work in concert with other transcription
the transcriptional profile of HeLa cells overexpressing                      factors to repress EVI127. CML (chronic myeloid leuke-
EVI1-FLAG or EVI1Δ324-FLAG was almost identical24.                           mia) blast crisis patient-derived cells express high EVI1
However, EVI1Δ324 does not replicate the transformative                      and β-catenin levels28. Knockdown of β-catenin or its
effects of EVI1 in rat fibroblasts, and is not known to have                  related co-transcription factor LEF1 (lymphoid enhancer
oncogenic activity nor is it linked currently with any                       factor 1) decreased EVI1 levels28. Bioinformatic analysis
myeloid malignancy24.                                                        indicated two potential tandem LEF1/β-catenin-binding
                                                                             sites present 1.44 kb upstream of EVI1, which are bound
EVI1 regulation                                                              by LEF1, as determined by ChIP assays28. Additional
Epigenetic regulation of EVI1                                                studies are needed to further clarify regulation of EVI1 by
   The region 5′ of EVI1 contains two CpG islands, one                       LEF1/β-catenin, RUNX1, GATA1, and/or ELK1.
close to the transcription start site of EVI1 and a second
located near MDS125. In an AML cell line that has low                        Post-translational modifications on EVI1
EVI1 expression, the CpG islands related to EVI1 and                           EVI1 has been reported to be phosphorylated at serine 196
MDS1 had a marked increase in methylation, suggesting                        (S196), S538, S858, and S86029,30. Stable isotope labeling of
that EVI1 expression can be regulated by methylation in                      amino acids followed by mass spectrometry (SILAC-MS)
AML cells25. Furthermore, AML cell lines with high EVI1                      identified EVI1-associated proteins. CK2 (casein kinase 2)
expression displayed active chromatin marks, with histone                    was confirmed to phosphorylate EVI1 residues S538 and
acetylation and enrichment of H3K4me3 (histone 3 lysine                      S858. Loss of phosphorylation was mediated by PP1α
4 tri-methylation) at the EVI1 promoter. In contrast cell                    (protein phosphatase 1 alpha), and it decreased DNA-
lines with low EVI1 expression have enrichment of the                        binding by the C-terminal ZF domain29. In contrast, phos-
repressive histone mark H3K27me3 (histone 3 lysine 27                        phorylation of S196 on the 6th zinc finger in the N-terminal
tri-methylation)25.                                                          ZF domain decreases DNA binding and repression by EVI1
                                                                             of promoters containing GATA-like motifs30. Although
EVI1 promoter                                                                phosphorylation of Ser858 and Ser860 did not affect EVI1
  The minimal promoter of EVI1 was localized to a 318                        DNA binding, loss of these phosphorylations blunted EVI1
nucleotide region 5′ of the EVI1 transcription start site                    transcriptional repression after cellular stress through
that does not contain a traditional TATA or CAAT box26                       reduced interaction of EVI1 with co-repressor CtBP131.
(Fig. 2). In the EVI1 minimal promoter, analysis of binding                  EVI1 is also acetylated by CBP (CREB binding protein or
motifs and site directed mutagenesis identified active                        KAT3A)/p300 (EP300, or KAT3B) and PCAF (P300/CBP-

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                  Page 4 of 14

associated factor or KAT2B)32. CBP-induced acetylation            EVI1 and shown to be required for EVI1 transformation
increased EVI1 transcriptional activity in luciferase assays32.   of rat fibroblasts40. This region was also critical for EVI1
In contrast, PCAF-mediated acetylation of EVI1 has been           repression of TGF-β (transforming growth factor beta)
reported to exhibit opposing effects on EVI1 activity. Co-        signaling and was thus termed the repressive domain (Rp)
expression of EVI1 with PCAF abrogated EVI1-mediated              (Fig. 1)41. Two consensus binding motifs for the tran-
Bcl-xL expression, suggesting that EVI1 acetylation blocked       scriptional co-repressor CtBP were identified in the EVI1
EVI1 transactivation activity at the Bcl-xL promoter33. In        Rp region. The PLDLS sequence at the residue 584 of
contrast, PCAF-mediated acetylation of K564 on EVI1               EVI1 is the major site of CtBP interaction7,8. Mutation of
increased its ability to transactivate GATA2, and this ability    the CtBP binding site at residue 584 abolished the ability
was lost in a K564A mutant that cannot be acetylated34.           of EVI1 to repress TGF-β-mediated growth arrest and
Overall, it is unclear whether these post-translational mod-      transformation of rat fibroblasts7,8.
ifications can occur simultaneously, or whether one mod-
ification can hinder the acquisition of another.                   Repression of other transcription factors by EVI1
                                                                    EVI1 can also directly bind several transcription factors
Transcriptional regulation by EVI1                                and inhibit their activity (Table 1B). EVI1 was able to
Transcriptional repression by EVI1                                repress GATA1-mediated activation of a synthetic pro-
  EVI1 co-immunoprecipitates with the H3K9me3                     moter. However, EVI1 does not bind to the canonical
methyltransferase SUV39H1 (suppressor of variegation              GATA1 motif42. Instead, EVI1 zinc fingers one and six
3–9 homolog 1) and the related H3K9me1/2 methyl-                  directly interact with the C-terminal zinc finger of
transferase G9a (euchromatic histone lysine methyl-               GATA1 in GST-fusion pull-down assays. Also, EVI1
transferase 2)35,36 (Table 1A). EVI1 and SUV39H1                  interaction with GATA1 decreased GATA1 DNA-binding
interaction required the N-terminal ZF domain of EVI1             ability. Mutation of EVI1 zinc fingers one and six abol-
and the C-terminal domain of SUV39H1. Histone                     ished GATA1 interaction and restored differentiation
methyltransferase assays showed SUV39H1 had methyl-               potential to 32Dcl3 cells in response to erythropoietin42.
transferase activity alone or in a complex with EVI1.               The 6th and 7th zinc finger of EVI1 was shown to
Furthermore, it was observed by the Nucifora and Delwel           directly interact with the C-terminal ETS-domain of PU.1
groups that EVI1-mediated repression of a GAL4 luci-              through co-immunoprecipitation and GST-fusion pull-
ferase construct was enhanced by SUV39H1 co-                      down assays. Binding of EVI1 to PU.1 did not prevent
expression35,36.                                                  DNA-binding ability of PU.1; instead it blocked associa-
  EVI1 represses PTEN (phosphatase and tensin homo-               tion of PU.1 with c-Jun (Jun Proto-Oncogene), a subunit
log) through its N-terminal ZF domain and via recruit-            of the transcription factor AP-1. Mutation of the 6th and
ment of the polycomb repressor complex 2 (PRC2),                  7th EVI1 zinc fingers mitigated EVI1 interaction with
including EZH2 (enhancer of zeste 2), by binding                  PU.1 and restored differentiation potential to 32Dcl3 cells
upstream of the PTEN transcription start site37. This             in response to G-CSF19. The 8th zinc finger in the C-
increased accumulation of the repressive H3K27me3                 terminal ZF domain of EVI1 was shown to interact with
mark and reduced histone acetylation at the PTEN locus            RUNX143. Binding of EVI1 repressed transcriptional
has been observed in human AML patient samples37.                 activity of RUNX1 by decreasing its DNA-binding43.
  EVI1 interacts through its N-terminal ZF domain with the        However, RUNX1 interaction with EVI1 had no effect on
de novo DNA methyltransferases DNMT3A and 3B38,39.                EVI1 DNA-binding. EVI1 interacts with the transcription
EVI1 expression correlated with differential hypermethyla-        factor SMAD3 through its N-terminal ZF domain41. EVI1
tion of over 200 genes, as compared to normal CD34+ cells,        interaction repressed SMAD3 activity leading to blocked
or to a previously reported DNA methylation profile in a           TGF-β mediated growth inhibition41.
separate cohort of 344 AML patients39. Unbiased motif
analysis of differentially methylated gene promoters showed       Transcriptional activation by EVI1
an enrichment of the motif recognized by the N-terminal             A number of gene targets are upregulated by EVI1
ZF domain of EVI139. DNMT3A was also found to be                  (Table 2). EVI1 interaction with histone acetyltransferases
highly expressed in EVI1-high AML samples compared to             has been reported to promote EVI1-mediated transcrip-
other AML subtypes. EVI1 expression levels correlated             tional activation32,34. EVI1 interaction with AP-1 subunits
positively with a stronger hypermethylation signature in          c-Fos and c-Jun was noticed as early as 1994 by Tanaka
AML patient samples39.                                            et al.44. EVI1-expressing cells exhibited increased c-Fos
                                                                  and c-Jun levels, and the C-terminal ZF domain of EVI1
Interaction with co-repressor CtBP                                was critical for activation of the c-Fos promoter44. Loss of
 A region just left to the C-terminal ZF domain of EVI1           EVI1 decreased c-Fos occupancy on the DNA, suggesting
was associated with transcriptional repression activity of        that EVI1 and AP-1 may act cooperatively at some loci6. A

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                                                           Page 5 of 14

Table 1 (A) EVI1 interactions with epigenetic regulators. (B) Biology of direct interaction of EVI1 with other
transcription factors.

(A)                                                           EVI1 interaction domain                                 Cellular models studied                        Ref

DNA methyltransferase
DNMT3A                                                        N-terminal zinc finger domain                            293T, SB1690CB                                 38,39

DNMT3B                                                        N-terminal zinc finger domain                            293T, SB1690CB                                 38,39

Histone methyltransferase
SUV39H1                                                       N-terminal zinc finger domain                            φE, 293T, HeLa                                 35,36

G9a                                                           N-terminal zinc finger domain                            φE, 293T, HeLa                                 35,36

EZH2                                                          N-terminal zinc finger domain                            THP-1, Jurkat, AML samples                     37

Histone acetyltransferase
                                                                                                                                                                     32
CBP                                                           Central region                                          Cos7
                                                                                                                                                                     32–34
PCAF                                                          N-terminal region/C-terminal region                     Cos7, HT-29, UCSD-AML1

(B)     TFs            Activity     EVI1 interaction domain                      Cellular models studied                        Biological outcome                     Ref

        Myeloid
         RUNX1         Down         8th zinc finger and central domain            NIH-3T3, 32Dcl3, 293T, K562                    Blocks myeloid differentiation         43

         GATA1         Down         1st and 6th zinc fingers                      32DEpo1, 32Dcl3, Cos7, AML14.3D10              Blocks myeloid differentiation         42

         PU.1          Down         6th and 7th zinc fingers                      32Dcl3 and 293T                                Blocks myeloid differentiation         19

        General
         SMAD3         Down         1st–7th zinc fingers                          32Dcl3                                         Blocks TGF-β responsiveness            41

(A) Epigenetic regulator proteins experimentally determined to interact with EVI1.
EVI1 ecotropic viral integration site 1, N-ter ZF domain N-terminal zinc finger domain, DNMT3A/B DNA methyltransferase 3A/B, SUV39H1 suppressor of variegation 3-9
homolog 1, G9a euchromatic histone lysine methyltransferase 2, EZH2 enhancer of zeste 2, CBP CREB binding protein a.k.a. KAT3A, PCAF P300/CBP-associated factor a.
k.a. KAT2B.
(B) Transcription factors experimentally determined to directly interact with EVI1, the interacting domain of EVI1 involved and the implications of the interaction on
the activity of the transcription factor.
TFs transcription factors, EVI1 ecotropic viral integration site 1, RUNX1 RUNX family transcription factor 1, GATA1 GATA binding protein 1, PU.1 transcription factor PU.1,
SMAD3 SMAD family member 3, NF-κB p65 nuclear factor kappa B family member p65.

SILAC-MS screen also confirmed c-Fos and c-Jun inter-                                    abnormalities, molecular alterations, pathological fea-
action with EVI129. This screen also identified several                                  tures, and poor prognosis46,50–52. In inv,(3) breaks most
additional transcription factors and co-factors that inter-                             frequently occur in a region between RPN1 (Ribophorin
act with EVI1, and 65% of EVI1-regulated genes were                                     1) and C3orf27, downstream of GATA2, that contains a
upregulated6,29. This highlighted the role of EVI1 as a                                 distal GATA2 hematopoietic enhancer (−77 kb, G2DHE)
transcriptional activator.                                                              and the region between C3orf50 and the first exon of the
                                                                                        MECOM locus that encodes for the MDS1-EVI1 tran-
EVI1 dysregulation in myeloid leukemia                                                  script (Fig. 3)53. The EVI1 and EVI1Δ324 transcripts
Chromosome 3 lesions leading to EVI1 overexpression                                     remain intact, but the MDS1-EVI1 transcript is frequently
  In the World Health Organization (WHO) classification                                  not expressed53. In t(3;3), the breakpoint frequently
of AML and related neoplasms, inversion or translocation                                occurs in between the MDS1 promoter and the first EVI1
of chromosome 3 at the MECOM locus [inv3(3;3)                                           exon, and MDS1-EVI1 transcript is frequently lost
(q21q26)/inv,(3) t(3;3)(q21;q26.2)/t(3;3)] have been                                    (Fig. 3)46. In 2014, the Delwel group and the Yamamoto
recognized as recurrent genetic abnormalities45 (Fig. 3).                               group identified that a new super enhancer of ~40 kb is
Inv(3)/t(3;3) is observed in ~1–2.5% of MDS and in a                                    formed from repositioning of the GATA2 distal hema-
similar percentage of AML patients46,47. Inv(3)/t(3;3)                                  topoietic enhancer that drives increased EVI1 expression
rearrangements can also be observed in up to 25–40% of                                  in inv(3)/t(3;3) AML53,54. The new enhancer region gen-
CML patients in blast crisis48,49. Despite their existence as                           erated by the chromosomal rearrangement was noted to
distinct clinical entities, MDS, AML, and CML with inv                                  contain a 9-kb region with a p300 binding site that
(3)/t(3;3) rearrangements have similar cytogenetic                                      interacts with the EVI1 promoter, and removal of this

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                                                     Page 6 of 14

Table 2 Transcriptional targets of EVI1.

Gene      Activity/levels Regulation                          Cellular models studied              Biological outcome                                           Ref

                                                                                                                                                                73
MYC       Up                 Transcriptional upregulation SKOV3, HeLa                              Active metabolism and apoptosis resistance
                                                                                                                                                                33
BcL-xL    Up                 Transcriptional upregulation HT-29, 293T                              Apoptosis resistance
                                                              AML samples
                                                                                                                                                                71
GPR56     Up                 Transcriptional upregulation UCSD-AML1, HNT-34                        Apoptosis resistance
                                                              AML samples
                                                                                                                                                                72
ITGA6     Up                 Transcriptional upregulation UCSD-AML1, HNT-34                        Apoptosis resistance
                                                              AML samples
                                                                                                                                                                44
c-Fos     Up                 Transcriptional upregulation P19, SKOV3, HeLa                         Activates AP-1
                                                                                                                                                                68
PBX1      Up                 Transcriptional upregulation HEL                                      Maintains AML stem cell phenotype
                                                              Primary murine BM
                                                                                                                                                                34
GATA2     Up                 Transcriptional upregulation EML-C1, HEL                              Maintains AML stem cell phenotype
                                                              Primary mouse EC
                                                                                                                                                                17,18
C/EBPα Down                  Transcriptional repression       32Dcl3, EML, DA-1, U937              Blocks differentiation
                                                                                                                                                                16
RUNX1     Down               Transcriptional repression       32Dcl3                               Blocks differentiation
                                                                                                                                                                37
PTEN      Down               Transcriptional repression       Primary murine BM AML                Activates metabolism and apoptosis resistance by
                                                              samples                              PI3K/AKT/mTOR pathway

Transcriptional targets of EVI1, the effect on the activity/levels of each target, and the biologic consequence of EVI1-mediated transcriptional regulation on the
target genes.
EVI1 ecotropic viral integration site 1, BM bone marrow, EC embryonic cells, PI3K phosphoinositide 3-kinase, AKT AKT serine/threonine kinase, mTOR mechanistic target
of rapamycin kinase, PTEN phosphatase and tensin homolog, MYC MYC proto-oncogene, BHLH transcription factor, BcL-xL BCL2 Like 1, GPR56 adhesion G protein-
coupled receptor G1, ITGA6 integrin subunit alpha 6, c-Fos Fos proto-oncogene, AP-1 transcription factor subunit, PBX1 PBX homeobox 1, GATA2 GATA binding protein
2, C/EBP CCAAT enhancer binding protein, CDK2 cyclin dependent kinase 2.

binding site attenuates EVI1 expression53. Transgenic                               interaction domain 1B), respectively55. Similar to the
mice in which the human inv(3) chromosomal abnorm-                                  “enhancer hijacking” of the GATA2 distal hematopoietic
ality was recapitulated through a bacterial artificial chro-                         enhancer in inv(3)/t(3;3), atypical 3q26 rearrangements
mosome developed leukemia, but if the relocated GATA2                               also seem to overexpress EVI1 through repurposing of
enhancer region was deleted, EVI1 expression declined                               enhancer elements from the translocation partners. In ten
and leukemia did not develop54. These seminal studies                               cases of atypical 3q26 rearrangements, EVI1 was over-
confirmed that EVI1 dysregulation in response to chro-                               expressed and the translocation partner whose enhancer
mosomal rearrangements in myeloid disease occurred not                              was repurposed had decreased expression, with the
from the typical generation of a fusion transcript, but                             exception of MYC in t(3;8)(q26;q24)55.
rather from “enhancer hijacking”53,54. This highlighted a
two-fold impact, one leading to overexpression of EVI1                              EVI1 fusion proteins
and the second causing haploinsufficiency of GATA2, as                                 Several translocations involving the MECOM locus do
it is no longer expressed in the rearranged chromosome53.                           result in the generation of fusion proteins. The two most
                                                                                    common being t(3;12)(q26;p13) and t(3;21)(q26;q24) that
Atypical 3q26 rearrangements                                                        result in ETS variant transcription factor 6 (also TEL)-
  In ~0.5–1% of AML and MDS patients, atypical chro-                                EVI1 and RUNX1-MDS1-EVI1 fusion proteins, respec-
mosome 3 rearrangements occur involving the MECOM                                   tively22,56 (Fig. 4). Both translocations are rare, found in
locus55. Most atypical 3q26 rearrangements have levels of                           less than 1% of myeloid malignancies56,57. Fluorescent
EVI1 overexpression comparable to inv(3)/t(3;3) cases,                              in situ hybridization demonstrated that the t(3;12)
similar phenotypic changes, and share a poor prog-                                  breakpoints are between the ETV6 exon 2 and 3 and on
nosis46,55. Atypical 3q26 rearrangements include, but are                           heterogeneous regions in 3q26, both 3′ and 5′ of MDS1 as
not limited to t(2;3)(q21;q26), t(3;7)(q26;q24), t(3;8)(q26;                        well as in between MDS1 and EVI156. The resulting
q24), and t(3;6)(q26;q25), which involve THADA                                      translocation fuses the first two exons of ETV6 with the
(THADA armadillo repeat containing), CDK6 (cyclin                                   entire MDS1-EVI1 or EVI1 transcript. Since no known
dependent kinase 6), MYC (V-Myc avian myelocytoma-                                  functional domain of ETV6 is added to EVI1 in the fusion
tosis viral oncogene homolog), and ARID1B (AT-rich                                  protein, it is thought that the oncogenic properties of the

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                                                                                              Page 7 of 14

                                                                                            Chromosome 3

                                           p24.3
                                           p24.2

                                                   p21.3

                                                                 p21.2
                                                                         p14.3
                                                                         p14.2

                                                                                                        q13.1

                                                                                                                q13.2
                                                                                                                q13.3

                                                                                                                                         q25.1
                                                                                                                                                 q26.1

                                                                                                                                                          q26.2
                                                                                                                                                          q26.3
                                                                                                q11.2
                                     p25

                                                   p23
                                                   p22

                                                                                  p13
                                                                                        p12

                                                                                                                        q21

                                                                                                                                q22
                                                                                                                                q23
                                                                                                                                q24

                                                                                                                                                          q27
                                                                                                                                                          q28
                                                                                                                                                          q29
                                                                                                p11
                         Chr3       Normal cells                                                                                                                                   Chr3
                         q21                                                                                                                                                       q26
                                    GATA2
                                                                                                                                                                        EVI1
                            GATA2
                                                                Chromatin                                                                                ~40 Mb
                                                                 Looping
                                           GATA2                                              Distal GATA2
                                                                                               Enhancer                              RPN1                                        EVI1
                                                                             ~128 kb

                         Chr3 inv(3)(q21;q26)                                                                                                                                     Chr3
                         q21                                                                                                                               EVI1      EVI1 EVI1    q26

                             GATA2
                                                                                                                                                         Inv(3) BP
                                                                                                                              Distal GATA2
                                           GATA2                                               RPN1                            Enhancer                                        EVI1
                                                                                                                                                              Chromatin
                                                                                                                                                               Looping

                         Chr3   t(3;3)(q21;q26)                                                                                                                                   Chr3
                         q21                                                                                                                                                      q26
                                                                            EVI1        EVI1 EVI1
                            GATA2
                                                                                                                 t(3;3) BP
                                                                                                                                     Distal GATA2
                                     GATA2                                                      EVI1                                                                       RPN1
                                                                                                                                      Enhancer

  Fig. 3 Schematic of the q21-26 locus on chromosome 3 in normal cells and cells with inv(3)(q21q26) or t(3;3)(q21q26). In both inv(3)
  (q21q26) or t(3;3)(q21q26), the breakpoints lead to juxtaposition of a region surrounding the distal GATA2 enhancer and the RPN1 gene in 3q21 with
  the EVI1 gene in 3q26. Breakpoints occur 3′ of the EVI1 gene in the inv(3)(q21q26) setting, whereas they occur 5′ of the EVI1 gene in the case of t(3;3)
  (q21q26). In both types of 3q21q26 rearrangement, the GATA2 enhancer induces EVI1 gene transcription instead of GATA2 expression and thus
  promotes leukemogenesis.

                                                                                                                                                                        Acidic
                       A                   EVI1                            NH2-         1       ZnF 2-7                                             Zn F 8- 1 0         Domain
                                                                                                                                                                                      -COOH

                                           MDS1

                                                             NH2-                                                                                                       Acidic
                                           MDS1-EVI1                                    1       Z n F 2- 7                                           ZnF 8-10                         -COOH
                                                                                                                                                                        Domain
                                                               PR Domain

                       B
                       RUNX1        NH2-           Runt Domain             TAD              -COOH

                       t(3:21)(q26;q24)
                       RUNX1-                                                                                                                                           Acidic
                       MDS1-EVI1 NH2-              Runt Domain                          1       ZnF 2-7                                             ZnF 8-10            Domain
                                                                                                                                                                                      -COOH

                       C
                                  ETV6                NH2-       HLH                                                           ETS               -COOH

                                  t(3:12)(q26;p13)
                                  ETV6-EVI1                         NH2-                                                                                                Acidic
                                                                                        1       ZnF 2-7                                           ZnF 8-10                            -COOH
                                                                                                                                                                        Domain

                                 ETV6-                                                                                                                                  Acidic
                                                      NH2-                              1       ZnF 2-7                                            ZnF 8-10                           -COOH
                                 MDS1-EVI1                                                                                                                              Domain

  Fig. 4 EVI1 and EVI1 fusion proteins. Schematic diagram of the EVI1, MDS1, MDS1-EVI1, RUNX1, RUNX1-MDS1-EVI1, ETV6, ETV6-EVI1, and ETV6-
  MDS1-EVI1 proteins.

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                               Page 8 of 14

fusion protein come from the inappropriate expression           7 in the dominate clone, suggesting that EVI1 could favor
and function of EVI1 driven by the ETV6 promoter56. In          expansion of clones with monosomy 7 or that EVI1 could
line with this, similar to other 3q26 rearrangements,           contribute to the genomic instability leading to monos-
myeloid malignancies expressing the ETV6-EVI1 fusion            omy 713.
are associated with dysmegakaryopoiesis and poor prog-
nosis58. The t(3;21) can generate RUNX1-MDS1-EVI1               Effect of EVI1 on hematopoietic stem cell proliferation/
fusion protein, where the DNA-binding RUNT domain of            differentiation
RUNX1 is fused to the whole MDS1-EVI1 protein (Fig.               EVI1 is known to directly interact with and repress the
4)59. Expression of the RUNX1-MDS1-EVI1 protein is              activity of a number of myeloid transcription factors
associated with disruption of RUNX1 and EVI1 regulatory         including GATA1, PU.1, and RUNX117,19,42,43. Enforced
networks. This is thought to be partly achieved by tran-        Evi1 expression transcriptionally repressed C/EBP-α in
scriptional repression of RUNX1 targets through                 murine hematopoietic cells18. EVI1-mediated repression
recruitment of co-repressors by EVI1 in the fusion pro-         of C/EBP-α was also observed in the murine hemato-
tein57. In mouse models of conditional RUNX1-MDS1-              poietic progenitor cell line 32Dcl317. Further confirma-
EVI1 expression or transplant models, the fusion protein        tion that EVI1 represses C/EBP family members is
is associated with development of hematopoietic dysplasia       needed through in vivo leukemia models and in patient-
and acute megakaryoblastic leukemia60.                          derived samples. EVI1 also regulates hematopoietic dif-
                                                                ferentiation and proliferation through transcriptional
EVI1 overexpression without chromosome 3 aberrations            repression of several miRNAs. EVI1 repressed miR-9
  Aberrant EVI1 expression can also occur in the absence        levels through binding to its regulatory region, recruiting
of chromosome 3 rearrangements. EVI1 overexpression is          DNMT3B, and inducing DNA methylation66. Decreased
observed in ~8–10% of MDS, 8% of de novo AML, and               miR-9 led to increased levels of its target genes FOXO1
30% of advanced CML, but it is unclear here how EVI1            and 3 (Forkhead Box O1 and 3)66. EVI1 expression was
overexpression occurs61. Several ChIP studies have shown        also found to decrease miR-449A levels, and ChIP ana-
that mixed lineage leukemia (MLL) and MLL fusion                lysis showed EVI1 bound miR-449A regulatory region67.
proteins, including MLL-AF9 and MLL-ENL bind to the             Repression of miR-449A by EVI1 increased expression of
EVI1 regulatory region, resulting in increased EVI1             the miR-449A-targets Notch1 and Bcl-2 in human AML
expression62,63. In a recent report in which MLL-AF9            cell lines67.
fusion gene was expressed either in murine Sca−Kit+               EVI1 transcriptionally activates the hematopoietic
(LSK) HSCs or in granulocyte monocyte precursors                proto-oncogene PBX1 (PBX homeobox 1) through bind-
(GMPs), LSK-MLL-AF9 cells had significantly higher               ing to its promoter region68. Knockdown of PBX1
levels of Evi1 than GMP-MLL-AF9 cells64. Additionally,          decreased EVI1-mediated transformation of primary
AMLs with high EVI1 expression have been shown to be            mouse bone marrow cells68. Comparing tissues from wild
associated with inferior relapse-free and overall survival65.   type to those from EVI1+/− and EVI1−/− mice, at
                                                                embryonic day 9.5, GATA2 expression was decreased in
Biologic consequences of 3q lesions and EVI1                    EVI1 depleted tissues69. EVI1 expression also correlated
overexpression                                                  with high expression of megakaryocytic markers, includ-
Genomic instability                                             ing the thrombopoietin receptor MPL70. Furthermore, in
  Utilizing SILAC-MS studies to determine EVI1 inter-           a mouse model of EVI1 leukemia, thrombopoietin
action partners, Bard-Chapeau et al. observed enrichment        expression correlated with EVI1 expression, and double
in protein domains associated with DNA repair, chro-            positive EVI1-thrombopoietin cells had enhanced sec-
matin remodeling, and transcription29. Furthermore, the         ondary leukemia formation ability in a serial bone marrow
EVI1 N-terminal ZF domain binds to GATA-like SSRs,              transplant assay70. Collectively, in myeloid malignancies
and EVI1 ChIP analysis revealed an increase in recombi-         expressing EVI1, transcriptional alterations of specific
nation rates near EVI1 bound SSR13,24. How EVI1                 myeloid transcription factors, and of miRNAs, contribute
increases genomic instability is not well characterized         to myeloid dysplasia.
beyond its protein interactions. However, a gene therapy
study using a Maloney murine leukemia virus vector to           Increased drug resistance
express NADPH-oxidase conducted in two patients to                Several pathways have been implicated in EVI1-
treat chronic granulomatous disease unfortunately caused        mediated resistance to apoptosis leading to drug-
integration of the vector at the MECOM locus. The               resistance. High EVI1 expression correlated with high
patients developed clonal expansion of myeloid cells            expression of the anti-apoptotic Bcl-xL protein in CML
bearing activating insertions in the MECOM locus and            patient samples33. Conversely, knockdown of EVI1 was
EVI1 overexpression. Both patients developed monosomy           shown to decrease Bcl-xL levels by approximately five

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                                                   Page 9 of 14

Table 3 Retrospective analysis of clinical outcome of patients with 3q26 genetic lesions.

Year       First author        N       CR (%)        Median OS (m)           1-year OS        Long term OS           Long term relapse probability             Ref

                                                                                                                                                               46
2010       Lugthart            79      31%           10.3                    N.D.             5-year OS: 5.7%        5-year RFS: 4.3%
                                                                                                                                                               75
2010       Grimwade            69      36%           N.D.                    N.D.             10-year OS: 3%         10-year CIR: 89%
                                                                                                                                                               45
2011       Sun                 30      42%           8.9                     33%              5-year OS: 3%          N.D.
                                                                                                                                                               87
2015       Wanquet             40      29%           10.6                    N.D.             4-year OS: 3%          N.D.
                                                                                                                                                               76
2020       Sitges, M           61      29%           8.4                     42%              4-year OS: 13%         N.D.

N number, ORR overall response rate, CR complete remission, OS overall survival, m months, RFS relapse-free survival, CIR cumulative incidence of relapse, N.D. not
discussed.

fold33. EVI1 interactions with the microenvironment are                             poor response to therapy, and has been linked with
also implicated in apoptosis-resistance. The adhesion                               acquisition of resistance to tyrosine kinase inhibitors48.
molecules ITGA6 (integrin subunit alpha 6) and GPR56
(adhesion G protein-coupled receptor G1) are highly                                 Monosomy 7 and MLL translocations
expressed in EVI1-positive AML, and their knockdown                                   Loss of one copy of chromosome 7 (monosomy 7, −7)
leads to increased apoptosis in response to Ara-C treat-                            or deletion of the long arm of chromosome 7 (−7q) is
ment and loss of RhoA (ras homolog family member A)                                 observed in 30–70% of MDS and AML with inv(3)/t
signaling, respectively71,72. In AML, cells high EVI1                               (3;3)74. Retrospective studies have shown that inv(3)/t(3;3)
expression correlated with high MYC and BCL2 expres-                                MDS/AML with −7/−7q display worse prognosis than
sion, with poorer clinical outcome73.                                               inv(3)/t(3;3) alone74,77. Which genetic alteration occurs
                                                                                    first is unclear, and likely varies on a case-by-case basis,
Clinical phenotypes and outcome of EVI1-positive                                    given the heterogeneity of the myeloid malignancies. As
myeloid malignancies                                                                noted above, in two cases where gene therapy activated
  MDS with EVI1 overexpression is commonly associated                               EVI1 expression through retroviral insertion, both cases
with dyserythropoiesis and with the presence of micro                               developed monosomy 7 in the dominant leukemic clone,
megakaryocytes51. Categorized as high risk, more than                               suggesting that EVI1 at least favors events leading to
half of inv(3)/t(3;3) MDS patients with EVI1 over-                                  monosomy 713. The q arm of chromosome 7 contains
expression progress to AML within ~2 years of diag-                                 several key genes whose haploinsufficiency is considered
nosis46,47. Furthermore, EVI1 overexpression correlates                             to be a loss of tumor-suppressor and thus contribute to
with shorter overall survival and poorer response to                                leukemia transformation. These genes include EZH2 and
treatment51. Overall survival of patients with EVI1-                                MLL3, as well as the cytoplasmic cellular regulators
positive MDS ranges from 13 to 17 months after diag-                                SAMD9 (Sterile Alpha Motif Domain Containing 9) and
nosis46,47. Like EVI1-positive MDS, AML with EVI1                                   SAMD9L78. Perhaps due to the ability of MLL and MLL-
overexpression often presents with myeloid dysplasia,                               fusion proteins to upregulate EVI1 transcription, EVI1
particularly of the erythrocyte and megakaryocytic linea-                           overexpression can be observed in ~30% of cases with
ges46,51. Studies have also reported EVI1 expression as an                          MLL translocation, and here EVI1 expression correlates
independent prognostic factor for poorer overall survival                           with poor prognosis63,65.
in AML, and high EVI1 expression is associated with
poorer response to therapy46,74,75. Several clinical studies                        Transcription factor mutations
have reported that, in 3q26-rearranged AML, the median                                Approximately 20% of MDS and AML patients with inv
overall survival after diagnosis remains approximately less                         (3)/t(3;3) express mutations in RUNX179. Another tran-
than 1 year, whereas long-term overall survival is less than                        scription factor IKZF1 (IKAROS family zinc finger) is also
15% (Table 3)47,74,76.                                                              mutated in up to 25% of cases of inv(3)/t(3;3) MDS or
  EVI1-expressing CML may also be associated with                                   AML. Since IKZF1 is located on chromosome 7, IKZF1
megakaryocytic dysplasia51. EVI1 expression is rarely                               mutations occur in clones without chromosome 7 dele-
detected in the chronic phase of CML, but is readily                                tions77. Although not a mutation, almost all MDS, AML,
detected in a significant proportion (25–40%) of blast                               and CML with inv(3)/t(3;3) have GATA2 haploinsuffi-
crisis of CML, suggesting that acquisition of EVI1                                  ciency due to the re-location of the GATA2 distal
expression can drive progression into blast crisis48,49.                            hematopoietic enhancer53,54. This was shown to con-
EVI1 expression in CML blast crisis is correlated with                              tribute to EVI1-driven leukemia transformation21. Of

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                              Page 10 of 14

note, despite loss of expression from one allele of GATA2,    or t(3;21), therapy with a second generation tyrosine
15% of inv(3)/t(3;3) can carry additional mutations in        kinase inhibitors and/or chemotherapy is utilized. EVI1-
GATA2 on the non-rearranged allele52,77.                      positive myeloid malignancies have been documented to
                                                              be relatively refractory to current therapies. There is no
Activating mutations in signaling pathways                    statistical difference in the overall 5-year survival rates
  A significant proportion of inv(3)/t(3;3) MDS and AML        between MDS and AML with inv(3)/t(3;3), which avera-
cases have activating mutations in RAS GTPase family          ges at 3–5%46,47,51,74. In a study by the Delwel group,
member (NRAS or KRAS), or in other RAS-signaling              allogenic hematopoietic stem cell transplant following the
pathway proteins, including PTPN11 (protein tyrosine          first clinical remission yielded increased survival odds in
phosphatase non-receptor type 11), and NF1 (neurofi-           AML patients with MLL translocation with EVI1 over-
bromin 1), which promote dysregulated RAS signaling           expression65. However, greater than 40% of the patients
and uncontrolled proliferation52,77,79. These mutations are   still succumbed to their disease65. Overall survival rate of
observed in 66–98% of inv(3)/t(3;3) MDS/AML52,77,79. A        patients with CML who initially respond but later pro-
greater percentage of AML cases with inv(3)/t(3;3) AML        gress on TKI therapy and acquire EVI1 overexpression is
carried RAS family mutations, as compared to the MDS          half compared to those without EVI1 expression48.
cases79.
                                                              Potential targeted therapies for EVI1-positive myeloid
Mutations in epigenetic machinery                             malignancies
  Low frequency of mutations in DNMT3, TET2 (tet                To date, following treatment of myeloid malignancies
methylcytosine dioxygenase 2), and IDH1/2 (isocitrate         with inv(3)/t(3;3) or EVI1 overexpression with targeted
dehydrogenase 1/2) were observed in AML or MDS with           therapies, including DNA hypomethylating drugs, vene-
inv(3)/t(3;3)46,51,77. However, mutations in the polycomb     toclax or glasdegib, or with FLT3 TKI or IDH1/2 inhibi-
group protein ASXL1 (ASXL transcriptional regulator 1)        tors, clinical outcome data are unavailable80. A targeted
were reported in ~20% of AML cases with inv(3)/t(3;3)77.      agent has yet to be identified and developed that exhibits
Mutations in splicing factors SF3B1 (splicing factor 3b       clinical efficacy specifically against EVI1-overexpressing
subunit 1) and U2AF1 (U2 small nuclear RNA auxiliary          myeloid malignancies.
factor 1) were also found in ~30–60% of inv(3)/t(3;3)
MDS or AML cases52,77. The biologic impact of these           Treatment with ‘epimodifiers’
‘epimutations’ in myeloid malignancies on the transcrip-        One promising target is the chromatin reader protein
tional signature attributed to inv(3)/t(3;3) and EVI1         BRD4 (bromodomain containing 4), which is involved in
overexpression remains to be elucidated.                      transcriptional activation, especially via sustaining the
                                                              activity of super enhancers, such as those of MYC, CDK4/
Mutations inversely correlated with EVI1                      6 and BCL2/Bcl-xL81. By also inhibiting GATA2 super
  Mutations in NPM1 (nucleophosmin 1) and C/EBP-α             enhancer, treatment with BET (bromodomain and extra-
inversely correlate with inv(3)/t(3;3) and EVI1 expres-       terminal motif) inhibitor could repress EVI1, as well as
sion55,77,79. Why EVI1 overexpression is not seen with        reverse EVI1-dependent transcriptional programs
NPM1 or C/EBP-α mutations is unknown. One possibility         through inactivating enhancers and super enhancers of
could be that survival of the clones with high EVI1           the key oncogenes (Fig. 5). Preclinical use of BET inhibitor
expression in combination with a NPM1 or C/EBP-α              treatment was reported to inhibit growth and induce
mutation is impaired.                                         apoptosis of an EVI1-overexpressing AML cell line82.
                                                              Since several BET inhibitors are already undergoing
Clinical outcome with standard therapy of                     clinical evaluation, they represent an attractive therapy
myeloid malignancies with inv(3)/t(3;3)                       option for myeloid malignancies with inv(3)/t(3;3) and/or
  Standard front-line treatment of MDS includes DNA           EVI1 overexpression53.
de-methylating agents like azacitidine and decitabine. In       Several transcriptional regulators including EVI1 are
advanced, high-risk MDS carrying inv(3)/t(3;3) with an        acetylated by CBP/p30032. For example, RUNX1 interacts
increased percent of bone marrow blasts between 5 and         with EVI1 and is positively regulated by acetylation32,83,84.
20%, or in overt transformation of MDS to secondary           Furthermore, the GATA2 distal enhancer that is relocated
AML (sAML), chemotherapy with cytarabine (Ara-C) and          and transactivates EVI1 in inv(3) and t(3;3) chromosomal
the anthracyclines, idarubicin and daunorubicin is com-       aberrations contains a p300 binding site that is critical in
monly employed. In CML, treatment in the chronic phase        driving EVI1 expression54. Additionally, the GATA2
generally begins with a tyrosine kinase inhibitor (TKI)       enhancer has increased read-through of enhancer RNAs
such as imatinib, dasatinib or nilotinib. In transformation   (eRNAs) at the breakpoints that cause its repositioning,
of CML into blast crisis (CML-BC) carrying inv(3)/t(3;3)      and the CBP/p300 inhibitor GNE-049 is reported to

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                          Page 11 of 14

Targeting EVI1 Transcription and Activity in MDS/AML with BET Inhibitors
                                                                            anti-apoptotic Bcl-xL suggests that BH3-mimetic inhi-
                          Hijacked GATA2                                    bitor targeting this anti-apoptotic protein could also have
                              enhancer                                      therapeutic value, alone or in combinations against EVI1-
     BET                                                                    expressing myeloid malignancies. Recently, the small
   inhibitor
                           EVI1 promoter                                    molecule compound pyrrole-imidazole polyamide, which
                                                                            inhibits the DNA-binding activity of N-terminal ZF
                                                                            domain of EVI1, was shown to induce apoptosis of EVI1-
                                                      Immune
                                                                            expressing AML cells due to downregulation of the EVI1
    Metabolism                  EVI1                surveillance            target GRP56, which was linked to EVI1-mediated resis-
     c-Myc                                           NF-κB p65              tance to apoptosis71. Several therapies targeting anti-
     AKT/mTOR
                                                                            apoptotic proteins are in clinical trials, and these agents
      Stem phenotype
                                               Environmental                could potentially be employed in treatment of EVI1-
                                                interaction
          Pbx1                                     GPR56
                                                                            expressing myeloid malignancies.
                              Growth/
          GATA2               Survival             ITGA6
                                                   SMAD3
          GATA1
          RUNX1
                               c-Myc                                        Targeting β-catenin-TCF7L2 activity
                               AKT/mTOR
          C/EBPα               c-Fos                                          In CML with inv(3)/t(3;3), β-catenin/TCF1 (T cell factor
          C/EBPε               Bcl-xL
                               GPR56
                                                                            1) signaling was reported to be activated, which positively
                               ITGA6                                        regulated EVI128. Therefore, along with treatment with
                               SMAD3
                                                                            BCR-ABL1 targeted TKI inhibition of β-catenin/TCF
  Fig. 5 Effects of targeting EVI1 transcription and activity in MDS/       signaling may be a promising strategy. Additionally, sev-
  AML with BET inhibitors. In AML cells with inv(3)(q21q26) or t(3;3)       eral groups have demonstrated that resistance to BET
  (q21q26), the hijacked GATA2 enhancer interacts with the EVI1
  promoter resulting in high expression of EVI. This leads to aberrant
                                                                            inhibitors in AML is mediated by the activity of β-catenin/
  regulation of multiple transcriptional programs including metabolism,     TCF7L2/c-Myc axis, resulting in re-expression of c-Myc
  stem cell phenotype, growth/survival, environmental interactions, and     despite treatment with BET inhibitor89. In this setting
  immune surveillance in AML cells. Treatment with BET inhibitors that      also, treatment with a β-catenin/TCF signaling inhibitor,
  evict BET proteins such as BRD4 from the chromatin of the GATA2           e.g., BC2059 (tegavivint), may not only reverse BET
  enhancer leads to downregulation of EVI1 and its activity in AML cells.
                                                                            inhibitor resistance but also exhibit synergy with BET
                                                                            inhibitor against AML with inv(3)/t(3;3) and/or EVI1
                                                                            overexpression90.
particularly inhibit eRNAs53,85. Therefore, targeted com-
bination therapy with this HAT inhibitor would simulta-                     Summary and future directions
neously cause deacetylation of the transcription factors,                     While great strides have been made, there is still much
leading to decreased transcriptional activity, as well as                   more to be elucidated regarding EVI1 biology and its
cause the loss of super enhancer function, thereby effec-                   contributions to leukemogenesis. EVI1 is a transcriptional
tively shutting down EVI1 transcriptional program.                          regulator that promotes a stem-like expression program in
  EVI1 can repress transcription through recruitment of                     hematopoietic progenitors, crucial to their self-renewal,
DNMT3A or B resulting in de novo methylation39,86.                          growth, and repopulating potential. The role of co-
EVI1 expression is also associated with hypermethylation                    expression of alternative EVI1 transcripts in myeloid
of over 200 genes in AML samples39,86. Consistent with                      malignancies remains to be determined. EVI1 regulates
this, DNA methyltransferase inhibitors have exhibited                       transcription through recruitment of epigenetic modifiers.
clinical activity in EVI1-overexpressing AML87. However,                    Recent studies have highlighted the dysregulated tran-
since monotherapy with DNA hypomethylating agent as a                       scriptome and signaling pathways that underpin the
first-line treatment for MDS exhibits only a modest                          aberrant biology, aggressive phenotype, and refractoriness
clinical efficacy, combination with other targeted thera-                    to standard therapy of EVI1-overexpressing myeloid
pies is likely to achieve superior efficacy against EVI1-                    malignancies (Fig. 5). How commonly the co-occurring
expressing MDS. Use of EZH2 inhibitor to abrogate                           genetic alterations and mutations, e.g., monosomy 7 and
dependency of EVI1-expressing MDS or AML with                               RAS pathway mutations, and their order of acquisition,
monosomy 7 on the residual normal EZH2 function may                         contribute to the aggressive phenotype and therapy-
exert added efficacy.                                                        refractoriness of EVI1-overexpressing myeloid malig-
                                                                            nancies has yet to be fully characterized. New probes or
Targeted therapy with BH3-mimetic apoptosis inducers                        alternative methodology need to be developed that will
  Recently, co-treatment with venetoclax, a BH3-mimetic                     assist in probing the clonal architecture of the co-
inhibitor of Bcl-2 protein, with azacitidine was approved                   mutations that occur with EVI1 dysregulation at the
for therapy of AML88. EVI1-mediated upregulation of the                     single-cell level. Importantly, functional genomic studies

Blood Cancer Journal
Birdwell et al. Blood Cancer Journal (2021)11:64                                                                                                             Page 12 of 14

need to be conducted to identify specific dependences that                                 7. Izutsu, K. et al. The corepressor CtBP interacts with Evi-1 to repress trans-
can be targeted to achieve superior efficacy against EVI1-                                    forming growth factor beta signaling. Blood. 97, 2815–2822 (2001).
                                                                                          8. Palmer, S. et al. Evi-1 transforming and repressor activities are mediated by
overexpressing myeloid malignancies. Large scale screens                                     CtBP co-repressor proteins. J. Biol. Chem. 276, 25834–25840 (2001).
with or without the presence of a promising therapeutic                                   9. Hoyt, P. R. et al. The Evi1 proto-oncogene is required at midgestation for
agent, e.g., by CRISPR technology, could also potentially                                    neural, heart, and paraxial mesenchyme development. Mech. Dev. 65, 55–70
                                                                                             (1997).
yield new knowledge for designing effective combination                                  10. Zhang, Y. et al. PR-domain-containing Mds1-Evi1 is critical for long-term
therapies. Analysis of the 3D chromatin architecture of                                      hematopoietic stem cell function. Blood. 118, 3853–3861 (2011).
“hijacked enhancers” at EVI1 locus and their response to                                 11. Kataoka, K. et al. Evi1 is essential for hematopoietic stem cell self-renewal, and
                                                                                             its expression marks hematopoietic cells with long-term multilineage repo-
treatment may also provide insights into effective ways to                                   pulating activity. J. Exp. Med. 208, 2403–2416 (2011).
decrease EVI1 expression, suggest promising treatments,                                  12. Du, Y., Jenkins, N. A. & Copeland, N. G. Insertional mutagenesis identifies genes
and potential mechanisms of resistance. How dysregulated                                     that promote the immortalization of primary bone marrow progenitor cells.
                                                                                             Blood. 106, 3932–3939 (2005).
EVI1 expression in myeloid malignancies creates immune                                   13. Stein, S. et al. Genomic instability and myelodysplasia with monosomy 7
evasion and T cell exhaustion also remains to be fully                                       consequent to EVI1 activation after gene therapy for chronic granulomatous
elucidated. In this exciting era of novel immunotherapies                                    disease. Nat. Med. 16, 198–204 (2010).
                                                                                         14. Kustikova, O. S. et al. Activation of Evi1 inhibits cell cycle progression and
new research avenues and potential strategies have already                                   differentiation of hematopoietic progenitor cells. Leukemia. 27, 1127–1138
been illuminated. These are likely to involve harnessing of                                  (2013).
the innate or adaptive immune mechanisms to overcome                                     15. Steinleitner, K. et al. EVI1 and MDS1/EVI1 expression during primary human
                                                                                             hematopoietic progenitor cell differentiation into various myeloid lineages.
immune tolerance or T cell exhaustion in eliminating                                         Anticancer Res. 32, 4883–4889 (2012).
myeloid malignancies, including those driven by EVI1                                     16. Morishita, K., Parganas, E., Matsugi, T. & Ihle, J. N. Expression of the Evi-1 zinc
dysregulation.                                                                               finger gene in 32Dc13 myeloid cells blocks granulocytic differentiation in
                                                                                             response to granulocyte colony-stimulating factor. Mol. Cell. Biol. 12, 183–189
                                                                                             (1992).
Conflict of interest
                                                                                         17. Kreider, B. L., Orkin, S. H. & Ihle, J. N. Loss of erythropoietin responsiveness in
T.M.K. receives research funding from BMS/Celgene, Amgen, AstraZaneca,
                                                                                             erythroid progenitors due to expression of the Evi-1 myeloid-transforming
Astellas, Pfizer, AbbVie, Genentech, JAZZ, Cellenkos, InCyte, and Ascentage; T.
                                                                                             gene. Proc. Natl Acad. Sci. USA 90, 6454–6458 (1993).
M.K. serves as a consultant/advisory board member for Agios, Pfizer, Abbvie,
                                                                                         18. Wilson, M. et al. EVI1 Interferes with Myeloid Maturation via Transcriptional
Genentech, JAZZ, Daiichi Sankyo and Novartis. C.D.D. receives research funding
                                                                                             Repression of Cebpa, via Binding to Two Far Downstream Regulatory Ele-
from Abbvie, Agios, Calithera, Cleave, BMS/Celgene, Daiichi-Sankyo,
                                                                                             ments. J. Biol. Chem. 291, 13591–13607 (2016).
ImmuneOnc and Loxo. C.D.D. serves as a consultant to Abbvie, Agios, Celgene/
                                                                                         19. Laricchia-Robbio, L., Premanand, K., Rinaldi, C. R. & Nucifora, G. EVI1 Impairs
BMS, Daiichi Sankyo, ImmuneOnc, Novartis, and Takeda. C.D.D. serves on an
                                                                                             myelopoiesis by deregulation of PU.1 function. Cancer Res. 69, 1633–1642
advisory board with stock options for Notable Labs. K.N.B. serves as a
                                                                                             (2009).
consultant to Iterion Therapeutics. All other authors have no conflict of interest
                                                                                         20. Shimizu, S. et al. EVI1 is expressed in megakaryocyte cell lineage and enforced
to declare.
                                                                                             expression of EVI1 in UT-7/GM cells induces megakaryocyte differentiation.
                                                                                             Biochem. Biophys. Res. Commun. 292, 609–616 (2002).
                                                                                         21. Yamaoka, A. et al. EVI1 and GATA2 misexpression induced by inv(3)(q21q26)
Publisher’s note                                                                             contribute to megakaryocyte-lineage skewing and leukemogenesis. Blood
Springer Nature remains neutral with regard to jurisdictional claims in                      Adv. 4, 1722–1736 (2020).
published maps and institutional affiliations.                                            22. Nucifora, G. et al. Consistent intergenic splicing and production of multiple
                                                                                             transcripts between AML1 at 21q22 and unrelated genes at 3q26 in (3;21)(q26;
Received: 7 January 2021 Revised: 25 February 2021 Accepted: 3 March                         q22) translocations. Proc. Natl Acad. Sci. USA 91, 4004–4008 (1994).
2021                                                                                     23. Pinheiro, I. et al. Prdm3 and Prdm16 are H3K9me1 methyltransferases required
                                                                                             for mammalian heterochromatin integrity. Cell. 150, 948–960 (2012).
                                                                                         24. Sayadi, A. et al. Functional features of EVI1 and EVI1Delta324 isoforms of
                                                                                             MECOM gene in genome-wide transcription regulation and oncogenicity.
References                                                                                   Oncogene. 35, 2311–2321 (2016).
 1. Mucenski, M. L. et al. Identification of a common ecotropic viral integration         25. Vazquez, I. et al. Down-regulation of EVI1 is associated with epigenetic
     site, Evi-1, in the DNA of AKXD murine myeloid tumors. Mol. Cell. Biol. 8,              alterations and good prognosis in patients with acute myeloid leukemia.
     301–308 (1988).                                                                         Haematologica. 96, 1448–1456 (2011).
 2. Morishita, K. et al. The human Evi-1 gene is located on chromosome 3q24-q28          26. Maicas, M. et al. Functional characterization of the promoter region of the
     but is not rearranged in three cases of acute nonlymphocytic leukemias                  human EVI1 gene in acute myeloid leukemia: RUNX1 and ELK1 directly reg-
     containing t(3;5)(q25;q34) translocations. Oncogene Res. 5, 221–231 (1990).             ulate its transcription. Oncogene. 32, 2069–2078 (2013).
 3. Fears, S. et al. Intergenic splicing of MDS1 and EVI1 occurs in normal tissues as    27. Maicas, M. et al. The MDS and EVI1 complex locus (MECOM) isoforms regulate
     well as in myeloid leukemia and produces a new member of the PR domain                  their own transcription and have different roles in the transformation of
     family. Proc. Natl Acad. Sci. USA 93, 1642–1647 (1996).                                 hematopoietic stem and progenitor cells. Biochim. Biophys. Acta. Gene Regul.
 4. Bordereaux, D., Fichelson, S., Tambourin, P. & Gisselbrecht, S. Alternative spli-        Mech. 1860, 721–729 (2017).
     cing of the Evi-1 zinc finger gene generates mRNAs which differ by the               28. Manachai, N. et al. Activation of EVI1 transcription by the LEF1/beta-catenin
     number of zinc finger motifs. Oncogene. 5, 925–927 (1990).                               complex with p53-alteration in myeloid blast crisis of chronic myeloid leu-
 5. Perkins, A. S., Fishel, R., Jenkins, N. A. & Copeland, N. G. Evi-1, a murine zinc        kemia. Biochem. Biophys. Res. Commun. 482, 994–1000 (2017).
     finger proto-oncogene, encodes a sequence-specific DNA-binding protein.               29. Bard-Chapeau, E. A. et al. EVI1 oncoprotein interacts with a large and complex
     Mol. Cell. Biol. 11, 2665–2674 (1991).                                                  network of proteins and integrates signals through protein phosphorylation.
 6. Bard-Chapeau, E. A. et al. Ecotopic viral integration site 1 (EVI1) regulates            Proc. Natl Acad. Sci. USA 110, E2885–E2894 (2013).
     multiple cellular processes important for cancer and is a synergistic partner for   30. White, D. J. et al. Phosphorylation of the leukemic oncoprotein EVI1 on serine
     FOS protein in invasive tumors. Proc. Natl Acad. Sci. USA 109, 2168–2173                196 modulates DNA binding, transcriptional repression and transforming
     (2012).                                                                                 ability. PLoS ONE 8, e66510 (2013).

Blood Cancer Journal
You can also read